US20030099611A1 - Manipulation and detection of protein phosphatase 2c-pp2calpha - expression in tumor cells for cancer therapy, prevention and detection - Google Patents

Manipulation and detection of protein phosphatase 2c-pp2calpha - expression in tumor cells for cancer therapy, prevention and detection Download PDF

Info

Publication number
US20030099611A1
US20030099611A1 US09/029,479 US2947998A US2003099611A1 US 20030099611 A1 US20030099611 A1 US 20030099611A1 US 2947998 A US2947998 A US 2947998A US 2003099611 A1 US2003099611 A1 US 2003099611A1
Authority
US
United States
Prior art keywords
pp2cα
cells
vector
gene
gene product
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US09/029,479
Other languages
English (en)
Inventor
Sara Lavi
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Ramot at Tel Aviv University Ltd
Original Assignee
Ramot at Tel Aviv University Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Ramot at Tel Aviv University Ltd filed Critical Ramot at Tel Aviv University Ltd
Assigned to RAMOT-UNIVERSITY AUTHORITY FOR APPLIED RESEARCH AND INDUSTRIAL DEVELOPMENT LTD. reassignment RAMOT-UNIVERSITY AUTHORITY FOR APPLIED RESEARCH AND INDUSTRIAL DEVELOPMENT LTD. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: LAVI, SARA
Publication of US20030099611A1 publication Critical patent/US20030099611A1/en
Priority to US10/678,477 priority Critical patent/US20040197798A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/40Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against enzymes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/16Hydrolases (3) acting on ester bonds (3.1)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2750/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
    • C12N2750/00011Details
    • C12N2750/14011Parvoviridae
    • C12N2750/14111Dependovirus, e.g. adenoassociated viruses
    • C12N2750/14122New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/154Methylation markers
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/156Polymorphic or mutational markers
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/158Expression markers

Definitions

  • the present invention relates to detection and methods of treating cancer by utilizing the gene human type protein phosphatase 2C (PP2C ⁇ and PP2C ⁇ ) and gene products thereof and kits for the practice of the invention; preparing native and transgenic organisms in which the gene products encoded by the human PP2C ⁇ gene or its homolog in other species are produced, or the expression of the native PP2C ⁇ gene is modified or knocked out.
  • P2C ⁇ and PP2C ⁇ gene human type protein phosphatase 2C
  • Genes have now been identified that are involved in transformation such as Ras, Fos PDGF, erb-B, erb-B2, RET, c-myc, Bci-2, APC, NF-1, RB, p53, etc.
  • the genes fall into two broad categories proto-oncogenes and tumor suppressor genes.
  • Proto-oncogenes code for proteins that stimulate cell division and when mutated (oncogenes) cause stimulatory proteins to be overactive with the result that cells over proliferate.
  • Tumor suppressor genes code for proteins that suppress cell division. Mutations and/or aberrant regulation can cause these proteins to be inactivated thereby rendering the cells without proliferation restraint.
  • E2F and p53 and others can act as both oncogene and tumor suppressor gene when improperly expressed.
  • oncogenes and tumor suppressor genes are motifs which act as transcription factors and as protein kinase. The identification of these specific genes have disclosed some of how the cell life cycle progresses.
  • Gene amplification is one of the distinct abnormalities associated with malignant cells and transformed cell lines [see generally “Gene Amplification in Mammalian Cells, A comprehensive Guide. edited by R. E. Hellems, Marcel Dekker, Inc. for a review of amplification.] This phenomenon is part of the genetic instability characterizing neoplastic cells and occurs rarely in normal cells. Some oncogenes and tumor suppressor genes have been shown to be amplified such as Ras, Erb, p53 etc.
  • Phosphorylation of structural and regulatory proteins including oncogenes and tumor suppressor genes is a major intracellular control mechanism in eukaryotes [Wera and Hemmings, 1995; Cohen, 1989]. Protein phosphorylation and dephosphorylation is part of the regulatory cycle for signal transduction, cell cycle progression and transcriptional control. Protein kinases and protein phosphatases both have roles in the phosphorylation—dephosphorylation cycle, respectively. Mutations in the genes coding for these proteins can lead to failure of protein phosphorylation. For example, in yeast, mutations of a type 2C protein phosphatases lead to a defect in osmoregulation [Shiozaki and Russell, 1995].
  • pp2c is a protein serine/threonine phosphatase [Cohen 1989].
  • the pp2c family consists of two cytoplasmic isoenzymes in mammalian tissues [McGowan and Cohen, 1987] and at least three pp2c-like enzymes in yeast show the same enzymatic and biochemical properties.
  • the two mammalian isoenzymes are monomers but differ slightly in molecular mass (44 KDa and 42 KDa) and are designated pp2c ⁇ and pp2c ⁇ .
  • viruses are very specialized infectious agents that have evolved, in many cases, to elude host defense mechanisms.
  • Adeno associated viruses are members of the family of parvoviruses for which tumor suppressive properties have already been described in 1960 [for review see Rommelaere and Tattersal, 1990]. They are a group of small viruses, with a ssDNA genome of approximately 5000 nucleotides, characterized by identical palindromic termini of 154 bases.
  • the left part of the AAVDA3 genome encodes four multifunctional, overlapping, non-structural proteins (Rep78, Rep68, Rep52 and Rep40) which are translated from differentially spliced mRNA driven by the P5 and P19 promoters (Accession numbers J01901, M12405, M12468, M12469).
  • P5 and P19 promoters accesion numbers J01901, M12405, M12468, M12469.
  • VP1-VP3 overlapping capsid polypeptides
  • These extremely small DNA viruses are represented in vertebrates by two genera, the autonomously replicating and the helper dependent parvovirus [Siegl et al., 1985].
  • helper-dependent adeno-associated viruses depend for their replication on coinfecting helper virus [Young and Mayor, 1979a,b], or on conditions of genotoxic stress [Yakobson et al., 1987] and comprise agents infecting humans without apparent disease [Cukor et al., 1984].
  • Helper viruses are adenoviruses [Atchison et al., 1965], herpes group viruses [Salo and Mayor, 1979] and vaccinia virus [Schlehofer et al., 1986].
  • the helper viruses share the ability to induce chromosomal damage early in their infection cycle [Schlehofer andzzy Hausen, 1982].
  • Tumor suppressive properties have been found for AAV [for review see Schlehofer, 1994]. It has been shown that the development of tumors induced in rodents by adenoviruses, herpes viruses or by transplantation of cells transformed by these viruses could be inhibited by infecting the animal cells with AAV [Kirschtein et al. 1968; Mayor et al., 1973; de la Maza and Carter, 1981; Ostrove et al., 1981]. The in vivo findings of tumor suppression are paralleled by results showing inhibition of cellular transformation in vitro. This could be shown for cells of different origin (hamster and mouse) transformed by viruses or by activated oncogenes.
  • a method and kit of detecting cancer in a patient by detecting alterations of the activity of the gene (PP2C ⁇ or PP2C ⁇ ) coding for human type protein phosphatase 2C (pp2c ⁇ ) and genetic polymorphisms thereof in a specimen isolated from the patient is disclosed.
  • the invention further provides a method of treating cancer including the steps of first determining the type of cancer and cells expressing the cancer and then preparing a vector which will specifically target the cancer cells and can include regulatory elements to control the expressibility of PP2C ⁇ .
  • the vector is then administered to the patient.
  • an antisense vector can be prepared.
  • the invention further provides a method of treating diseases due to aberrant phosphorylation due to alteration of expression of PP2C ⁇ by controlling PP2C ⁇ expression.
  • FIGS. 1 A-B are graphs of a FACS analysis of CO60 and two AAV/neo cell lines 913 and 916 as prepared for cell cycle analysis. 24 hours after seeding the cell were trypsinized and washed with PBS. The cells were resuspended in 1 ml buffer containing 0.1% sodium citrate, 0.1% triton X-100 and 50 ⁇ g propidium iodide, and then processed in the FACS.
  • FIG. 2 is a photograph of a Southern Blot Analysis showing CHINT is associated with AAV integration in different AAV/neo cell lines.
  • 93R is a revertant that lost the whole chromosome containing the AAV.
  • A6 is a mouse cell line.
  • FIG. 3 is a schematic representation of the organization of the integrated AAV and the flanking cellular sequences in 9-3 cells.
  • a genomic library was prepared from C9-3 cells using the EMBL-4 lambda phage and scored for AAV positive clones.
  • a clone of 13Kb- ⁇ SL9-1 was isolated and later subcloned to a blue-script vector. Plasmids pSL9-11 (13 Kb), pSL9-8 (10 Kb) and pSL9-6 (3 Kb) were obtained as indicated in the figure.
  • FIGS. 4 A-B wherein (A) is a photograph of a Southern Blot Analysis showing AAV is adjacent to the gene coding to PP2C ⁇ in 9-3 cells,
  • the CHINT and the PP2C ⁇ sequences are adjacent (4 Kb EcoRI fragment).
  • the AAV CHINT and PP2C ⁇ are in a close proximity in 9-3 cells (the 5.6 Kb XbaI fragment).
  • (S) pSL9-6 is adjacent to PP2C ⁇ in the wild type Chinese hamster cells.
  • FIGS. 5 A-C are photographs of a Southern blot analysis of DA3 (lane 8) and DA3J1-DA3J7 cells lines (lanes 1-7). Genomic DNA was digested with BglII. The blots were hybridized sequentially with an AAV/neo JDT277, pSL9-6 and PP2C ⁇ PCR probes. A 4 Kb fragment hybridized to the AAV probe and pSL9-6 probe in J3 (lane 3), J4 (lane 4) and J6 (lane 6). A fragment smaller than 4 Kb hybridized to both AAV and PP2C ⁇ probe in J1 (lane 1), J2 (lane 2), J5 (lane 5) and J6 (lane 6).
  • FIG. 6 is a photograph which shows the alteration in PP2C ⁇ mRNA in response to carcinogen treatment. Forty ⁇ g of total RNA were isolated from CO60 and C9-3 cells 48 hours after treatment with MNNG (7.5 ⁇ g/ml and 2.5 ⁇ g/ml respectively), and from untreated cells and fractionated on a denaturing gel (1.2% agarose/6.6% formaldehyde gel). The gel was blotted and hybridized consecutively with 32 P-labeled rat PP2C ⁇ cDNA (A) pSL9-1 DNA (B) and rRNA cDNA (C).
  • A 32 P-labeled rat PP2C ⁇ cDNA
  • B pSL9-1 DNA
  • C rRNA cDNA
  • FIG. 7 is a photograph which shows gel electrophoresis anaylsis after 25, 30 and 35 PCR cycles.
  • the 25 cycle PCR cycle for PP2C ⁇ cDNA in the normal and tumor tissues is not shown since a visible product was not found.
  • FIG. 8 is a photograph which shows gel electrophoresis anaylsis after 25, 30, 30 and 35 PCR cycles of aliquots of the oligo dT-primed cDNA obtained from CHE cell line, or from its adjacent transformed cell line (CO60), subjected to PCR reactions using the specific PP2C ⁇ and the ⁇ -actin sense and anti-sense primers.
  • FIGS. 9 A-B are schematic representations of plasmids that contain PP2C ⁇ cDNA in the (A) sense orientation (pYM001) and in the (B) antisense orientation (pYM002).
  • FIG. 10 is a photograph which shows gel electrophoresis anaylsis of immunoprecipitation of liver extracts with a panel of monoclonal antibodies raised against pp2c ⁇ ; 1D5, 2A3, 9F4, 9F1, are monoclonal antibodies used to precipitate pp2c ⁇ from liver extract; 801 and 351 are rabbit polyclonal antibodies used for detection after immunoblotting.
  • FIG. 11 is a schematic representation of a genomic ⁇ 100 clone containing the first translated exon of PP2C ⁇ .
  • the phage was cloned from a CHO library. The sequenced regions are indicated by cross hatching (SEQ ID Nos:15 and 16).
  • FIG. 12 is a photograph which shows gel electrophoresis wherein lane 1: Cotransfection with pSK1 and pAV2; lane 2: Transfection with the SV40 plasmid pSK1 SV40 replicates; lane 3: Cotransfection of pSVK1 and a plasmid harboring 140 bp from the AAV genome nucleotide 125-263; lane 4: Cotransfection of pSVK1 with pSL9-6.
  • FIG. 13 is a photograph of a Northern blot wherein RNA from various mouse tissues is hybridized with PP2C ⁇ cDNA demonstrating that there are several mRNAs of different sizes ranging from less than 2 kb to higher than 5.0 kb.
  • RNA was extracted from ovary (O), Testis (T), Kidney (K), Liver (L), Muscle (M), Heart (H), Lung (Lu) and Brain (B).
  • the present invention discloses a method of detecting cancer in a patient by detecting alterations in gene activity of the gene (PP2C ⁇ ) coding for human type protein phosphatase 2C (pp2c ⁇ ) and genetic polymorphisms thereof in a specimen isolated from the patient.
  • the gene activity of the patient is compared to that of normal controls.
  • Alterations in activity can be a down-regulation of the gene activity or conversely an up-regulation resulting in changes in phosphorylation.
  • alterations can result in aberrant function or absence of the gene product and in a change in distribution of the gene product within the cell itself.
  • Polymorphisms are variants in the gene sequence. They can be sequence shifts found between different ethnic and geographic locations which, while having a different sequence, produce functionally equivalent gene products, isoforms. Polymorphisms also encompass variations which can be classified as alleles and/or mutations which can produce gene products which may have an altered function. Polymorphisms also encompass variations which can be classified as alleles and/or mutations which either produce no gene product, an inactive gene product or increased levels of gene product. Polymorphisms as used herein can also encompass variations which are due to differences in DNA methylation in control and coding regions. Further, the term is also used interchangeably with allele as appropriate.
  • Cancer is defined as transformed or malignant cells, i.e. cells undergoing uncontrolled growth and spread (see generally, Scientific American September, 1996 for a review).
  • the present invention there is more than on form of the gene product of PP2C ⁇ and that one may be reduced or altered in cells while another specific form of PP2C ⁇ will be elevated or more prominent compared to normal controls.
  • the cells can be any cell type that shows alteration in PP2C ⁇ activity in a disease state.
  • a second gene may be controlled by the alteration in the activity of PP2C ⁇ such that their products are elevated or reduced and can be monitored by the method of the present invention. New transcripts, absence of transcripts or alterations in the protein coded by these transcripts are monitored.
  • pp2c ⁇ is itself phosphorylated as it has several phosphorylation sites including tyrosine, serine and thyronine and that failure to phosphorylate it properly will cause malfunction of the pp2c ⁇ protein. Further, pp2c ⁇ also dephosphorylates itself. A failure in its autophosphorylation will have effects on cell cycle regulation.
  • Samples can be biopsied material from suspected precancerous lesions or any tissue or bodily fluid which can be assayed for PP2C ⁇ activity or gene product as described herein. Bodily fluids such as blood, urine, cerebrospinal fluid and saliva can be examined as is appropriate.
  • the detection of PP2C ⁇ activity is by assaying the specimen for mRNA complementary to PP2C ⁇ DNA including polymorphisms thereof with an assay selected from the group consisting of in situ hybridization, Northern blotting and reverse transcriptase—polymerase chain reaction.
  • the detecting of PP2C ⁇ activity and cellular distribution is by assaying the specimen for a PP2C ⁇ gene product including polymorphisms and peptide fragments thereof with an assay selected from the group consisting immunohistochemical and immunocytochemical staining, ELISA, RIA, immunoblots, immunoprecipitation, Western blotting, functional assays for activity of gene product, assays for phosphorylation patterns and protein truncation test.
  • Target proteins which are dephosphorylated by pp2c ⁇ can have different size characteristics on PAGE and different isoelectric points as well as changes in function such as their ability to interact with other proteins, RNA, DNA and other cellular components.
  • the method of the present invention screens for the gene product in bodily fluids.
  • the level of gene product in the bodily fluid is affected as for example more can be released from the cell if glycosylation or signal sequences are affected. Incomplete protein fragments may result from interrupted translation which are then released from the cell and are monitored.
  • the present invention recognizes alternately spliced forms of the mRNA for pp2c ⁇ giving rise to different sizes and/or function in different tissues and assays are designed to recognize the alternately spliced forms in the appropriate tissues.
  • the identification of alterations in the gene product in a specific bodily fluid will indicate the source/location of a tumor. For example, with a tumor in the central nervous system, the gene product would be found in the cerebrospinal fluid. Similarly the location of other tumors or other diseases would determine which bodily fluids to screen and the converse as would be known to those skilled in the art.
  • the present invention also provides for a kit for detecting PP2C ⁇ activity and/or alteration either at the mRNA level or gene product level.
  • the kit includes molecular probes for mRNA for PP2C ⁇ mRNA and detection means for detecting the molecular probe and thereby the mRNA.
  • the kit can contain probes for detecting the PP2C ⁇ gene product.
  • the detecting means are in general are antibodies with high specificity for the gene product or agents which mimic natural proteins which bind to the PP2C ⁇ gene product other agents as known in the art may also be used.
  • the antibodies are made as described herein below and in Example 3, which specifically recognize the PP2C ⁇ or PP2 ⁇ gene products (including on the cell surface) including polymorphisms thereof, and detection means for detecting the binding of the antibody thereby indicating the presence of the gene product and also distinguishing one from the other.
  • kits can also contain antibodies directed against-secondary gene products that are affected by the alteration in function of the PP2C ⁇ gene.
  • the present invention discloses a method of detecting cancer in a patient by detecting altered levels of PP2C ⁇ gene activity compared to normal patients in a specimen isolated from a patient.
  • the present invention also provides for a kit for detecting PP2C ⁇ activity.
  • the kit includes molecular probes for mRNA for PP2C ⁇ polymorphisms thereof and detection means for detecting the molecular probe and thereby the mRNA or antibodies or other means of identifying alterations in the level of the gene product over normal controls as described herein.
  • the present invention provides an antibody, either polyclonal or monoclonal, which specifically binds to a polypeptide/protein encoded by the PP2C ⁇ gene as described in Example 3 herein below.
  • the antibodies of the present invention are used in identifying the gene product of PP2C ⁇ and PP2C ⁇ .
  • the present invention provides monoclonal and polyclonal antibodies raised against recombinantly produced PP2C ⁇ , NDDTDSASTD (SEQ ID No:1), YKNDDTDSTSTDDMW (SEQ ID No:2), recombinantly produced pp2c ⁇ and PNKDNDGGA (SEQ ID No:3).
  • the present invention also provides for isolated and purified peptides NDDTDSASTD (SEQ ID No:1), YKNDDTDSTSTDDMW (SEQ ID No:2) and PNKDNDGGA (SEQ ID No:3).
  • the peptides can be produced recombinantly.
  • the invention further provides antibodies that will recognize the special structures at the 5′UTR or the RNA-proteins complexes responsible for the controlled expression of PP2C ⁇ .
  • Antibody which recognizes specifically the special RNA structures is also provided.
  • the entire pp2c ⁇ protein or peptide sequences thereof can be used as an immunogen as well as polymorphisms thereof.
  • anti-idiotypic antibodies can be made against these antibodies.
  • the antibodies may be either monoclonal or polyclonal.
  • the antibodies may be prepared against a synthetic peptide based on the sequence, or prepared recombinantly by cloning techniques or the natural gene product and/or portions thereof may be isolated and used as the immunogen.
  • proteins or peptides can be used to produce antibodies by standard antibody production technology well known to those skilled in the art as described generally in Harlow and Lane, Antibodies: A Laboratory Manual , Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1988.
  • a host such as a rabbit or goat, is immunized with the protein or peptide, generally with an adjuvant and, if necessary, coupled to a carrier; antibodies to the protein are collected from the sera.
  • the technique involves hyperimmunization of an appropriate donor, generally a mouse, with the protein or peptide fragment and isolation of splenic antibody producing cells. These cells are fused to a cell having immortality, such as a myeloma cell, to provide a fused cell hybrid which has immortality and secretes the required antibody. The cells are then cultured, in bulk, and the monoclonal antibodies harvested from the culture media for use.
  • an appropriate donor generally a mouse
  • a cell having immortality such as a myeloma cell
  • the antibody can be bound to a solid support substrate or conjugated with a detectable moiety or be both bound and conjugated as is well known in the art. (For a general discussion of conjugation of fluorescent or enzymatic moieties see Johnstone and Thorpe, Immunochemistry in Practice , Blackwell Scientific Publications, Oxford, 1982.) The binding of antibodies to a solid support substrate is also well known in the art.
  • the detectable moieties contemplated with the present invention can include, but are not limited to, fluorescent, metallic, enzymatic and radioactive at markers such as biotin, gold, ferritin, alkaline phosphatase, ⁇ -galactosidase, peroxidase, urease, fluorescein, rhodamine, tritium, 14 C and iodination. Additionally, toxins can be coupled to the antibody for targeted delivery.
  • the present invention also provides for transgenic human PP2C ⁇ gene and polymorphic PP2C ⁇ gene, animal and cellular (cell lines) models as well as for knockout PP2C ⁇ models. These models are constructed using standard methods known in the art and as set forth in U.S. Pat. Nos.
  • the present invention provides vectors comprising an expression control sequence operatively linked to the nucleic acid sequence of the PP2C ⁇ gene and portions thereof as well as polymorphic sequences thereof (see Examples herein below).
  • the present invention further provides host cells, selected from suitable eucaryotic and procaryotic cells, which are transformed with these vectors.
  • the vectors can be introduced into cells or tissues by any one of a variety of known methods within the art. Such methods can be found generally described in Sambrook et al., Molecular Cloning: A Laboratory Manual, Cold Springs Harbor Laboratory, New York (1992), in Ausubel et al., Current Protocols in Molecular Biology , John Wiley and Sons, Baltimore, Md. (1989), Chang et al., Somatic Gene Therapy , CRC Press, Ann Arbor, Mich. (1995), Vega et al., Gene Targeting , CRC Press, Ann Arbor, Mich.
  • such vectors are known or can be constructed by those skilled in the art and should contain all expression elements necessary to achieve the desired transcription of the sequences.
  • Other beneficial characteristics can also be contained within the vectors such as mechanisms for recovery of the nucleic acids in a different form.
  • Phagemids are a specific example of such beneficial vectors because they can be used either as plasmids or as bacteriophage vectors. Examples (see Example herein below) of other vectors include viruses such as bacteriophages, baculoviruses and retroviruses, DNA viruses, cosmids, plasmids, liposomes and other recombination vectors.
  • the vectors can also contain elements for use in either procaryotic or eucaryotic host systems. One of ordinary skill in the art will know which host systems are compatible with a particular vector.
  • Recombinant methods known in the art can also be used to achieve the sense, antisense or triplex inhibition of a target nucleic acid.
  • vectors containing antisense nucleic acids can be employed to express protein or antisense message to reduce the expression of the target nucleic acid and therefore its activity.
  • ribozymes can be generated and used to “knock-out” the mRNA expression of the gene [Cech, 1986; Cech, 1990; Hampel et al, 1993; Sullivan, 1994].
  • a specific example of DNA viral vector for introducing and expressing recombinant sequences is the adenovirus derived vector Adenop53TK.
  • This vector expresses a herpes virus thymidine kinase (TK) gene for either positive or negative selection and an expression cassette for desired recombinant sequences.
  • TK herpes virus thymidine kinase
  • This vector can be used to infect cells that have an adenovirus receptor which includes most cancers of epithelial origin as well as others.
  • This vector as well as others that exhibit similar desired functions can be used to treat a mixed population of cells and can include, for example, an in vitro or ex vivo culture of cells, a tissue or a human subject.
  • Additional features can be added to the vector to ensure its safety and/or enhance its therapeutic efficacy.
  • Such features include, for example, markers that can be used to negatively select against cells infected with the recombinant virus.
  • An example of such a negative selection marker is the TK gene described above that confers sensitivity to the antibiotic gancyclovir. Negative selection is therefore a means by which infection can be controlled because it provides inducible suicide through the addition of antibiotic. Such protection ensures that if, for example, mutations arise that produce altered forms of the viral vector or recombinant sequence, cellular transformation will not occur.
  • Features that limit expression to particular cell types can also be included. Such features include, for example, promoter and regulatory elements that are specific for the desired cell type.
  • recombinant viral vectors are useful for in vivo expression of a desired nucleic acid because they offer advantages such as lateral infection and targeting specificity.
  • Lateral infection is inherent in the life cycle of, for example, retrovirus and is the process by which a single infected cell produces many progeny virions that bud off and infect neighboring cells. The result is that a large area becomes rapidly infected, most of which was not initially infected by the original viral particles. This is in contrast to vertical-type of infection in which the infectious agent spreads only through daughter progeny.
  • Viral vectors can also be produced that are unable to spread laterally. This characteristic can be useful if the desired purpose is to introduce a specified gene into only a localized number of targeted cells.
  • viruses are very specialized infectious agents that have evolved, in many cases, to elude host defense mechanisms. Typically, viruses infect and propagate in specific cell types.
  • the targeting specificity of viral vectors utilizes its natural specificity to specifically target predetermined cell types and thereby introduce a recombinant gene into the infected cell.
  • the vector to be used in the methods of the invention will depend on desired cell type to be targeted and will be known to those skilled in the art. For example, if breast cancer is to be treated then a vector specific for such epithelial cells would be used. Likewise, if diseases or pathological conditions of the hematopoietic system are to be treated, then a viral vector that is specific for blood cells and their precursors, preferably for the specific type of hematopoietic cell, would be used.
  • Retroviral vectors can be constructed to function either as infectious particles or to undergo only a single initial round of infection.
  • the genome of the virus is modified so that it maintains all the necessary genes, regulatory sequences and packaging signals to synthesize new viral proteins and RNA. Once these molecules are synthesized, the host cell packages the RNA into new viral particles which are capable of undergoing further rounds of infection.
  • the vector's genome is also engineered to encode and express the desired recombinant gene.
  • the vector genome is usually mutated to destroy the viral packaging signal that is required to encapsulate the RNA into viral particles. Without such a signal, any particles that are formed will not contain a genome and therefore cannot proceed through subsequent rounds of infection.
  • the specific type of vector will depend upon the intended application.
  • the actual vectors are also known and readily available within the art or can be constructed by one skilled in the art using well-known methodology.
  • the recomnbinant vector can be administered in several ways and in combination with a suitable pharmaceutical carrier. If viral vectors are used, for example, the procedure can take advantage of their target specificity and consequently, do not have to be administered locally at the diseased site. However, local administration can provide a quicker and more effective treatment, administration can also be performed by, for example, intravenous or subcutaneous injection into the subject. Injection of the viral vectors into a spinal fluid can also be used as a mode of administration, especially in the case of neuro-degenerative diseases. Following injection, the viral vectors will circulate until they recognize host cells with the appropriate target specificity for infection.
  • An alternate mode of administration of a PP2C ⁇ vector can be by direct inoculation locally at the site of the disease or pathological condition or by inoculation into the vascular system supplying the tumor with nutrients.
  • Local administration is advantageous because there is no dilution effect and, therefore, a smaller dose is required to achieve expression in a majority of the targeted cells. Additionally, local inoculation can alleviate the targeting requirement required with other forms of administration since a vector can be used that infects all cells in the inoculated area. If expression is desired in only a specific subset of cells within the inoculated area, then promoter and regulatory elements that are specific for the desired subset can be used to accomplish this goal.
  • non-targeting vectors can be, for example, viral vectors, viral genome, plasmids, phagemids and the like.
  • Transfection vehicles such as liposomes can also be used to introduce the non-viral vectors described above into recipient cells within the inoculated area. Such transfection vehicles are known by one skilled within the art.
  • a virus vector based on modified AAV is used.
  • AAV has been shown to integrate into the human genome in chromosome 19q13.3. Alteration of the AAV genome in a mode that will allow it to integrate in a site specific manner into the PP2C ⁇ regulatory region is used. (see Example 7)
  • the invention further provides a method of treating cancer including the steps of first determining the type of cancer and cells expressing the cancer and then preparing a vector as described herein above which will specifically target the cancer cells and includes regulatory elements to control the expressibility of PP2C ⁇ .
  • the vector is then administered to the patient and can include a suitable pharmaceutical carrier which will not affect bioactivity of the vector.
  • an antisense vector can be prepared and used to control the expression of PP2C ⁇ .
  • pp2c is a protein serine/threonine phosphatase [Cohen 1989]. It is unique among phosphatases since it requires magnesium and is not sensitive to certain phosphatase inhibitors such as okadaic acid [Cohen 1991].
  • the pp2c family consists of two cytoplasmic isoenzymes in mammalian tissues [McGowan and Cohen, 1987] and at least three pp2c-like enzymes in yeast show the same enzymatic and biochemical properties.
  • the two mammalian isoenzymes are monomers but differ slightly in molecular mass (44 KDa and 42 KDa) and are designated pp2c ⁇ and pp2c ⁇ .
  • a 106 kb cosmid coding for pp2c ⁇ and additional proteins FosB and ERCCI has been sequenced [Martin-Gallardo et al., 1992] (GENBANK accession number: M89651). Further, the cDNA sequences of PP2C ⁇ in humans is known [Mann et al, 1992]. However, attempts to align the 5′UTR of the cDNA with the genomic sequences were not successful.
  • UTR consists of several small exons with large introns and propose that PP2C ⁇ and FosB have a common regulatory region (ERCCI may also share the regulatory region).
  • PP2C ⁇ is a very large gene and that the 5′ end and the control region do not reside within the 106 kb cosmid in a region located 5′ to the 106 kb cosmid.
  • the region of 9 kb from the cosmid was not sequenced due to the high G/C content and it may contain the 5′UTR region and the promoter.
  • the AAV virus and/or CHINT or other regulatory sequences related to the PP2C ⁇ gene are used in the vector, particularly those which are used to treat patients.
  • CHINT is a cellular sequence which was recombined into the AAV in 9-3 cells; the sequence is set forth in Table 5 (int.li; SEQ ID No:19).
  • the vector can either integrate into the regulatory control of PP2C ⁇ and alter its expression in the same way as AAV alters cells into which it integrates as it is an oncosuppressive virus.
  • PP2C ⁇ has a very long 5′ and 3′ UTR (they are larger than the coding capacity). Specific folding of the RNA and interaction with specific sets of proteins might effect its expression dramatically. At certain stages there might different modes of folding and these different proteins may interact with the RNA and alter its expression.
  • the invention further provides a method of treating cancer by using an AAV based vector or other vector for cancer treatment that only functions specifically in cells in which PP2C ⁇ is improperly activated.
  • the vector is administered to those who have been diagnosed with a tumor as is known to those skilled in the art.
  • the AAV vector (or other regulatory factor as disclosed herein) in one embodiment is under the control of a promotor, rep, that is expressed in transformed cells.
  • the integrated vector will control PP2C ⁇ expression in the cell reversing transformation as shown in the Examples. Further, the vector will be targeted to the cell type that has been transformed.
  • Fab fragments and other means known in the art can be used to insure that the antibodies upon administration to a patient do not have secondary unwanted effects.
  • a ligand or other molecule which can specifically bind to the PP2C ⁇ gene product can be used. The present invention therefore provides a method of binding the gene product of PP2C ⁇ expressed on the surface of a cell to induce signal transduction thereby suppressing the transformed phenotype.
  • the invention further provides a method of treating diseases due to aberrant phosphorylation due to alteration of expression of PP2C ⁇ by controlling PP2C ⁇ expression.
  • diseases due to aberrant phosphorylation due to alteration of expression of PP2C ⁇ by controlling PP2C ⁇ expression can be neurologic.
  • behavioral changes could be associated with aberrant phosphorylation.
  • fosB and PP2C ⁇ are on the 106 kD cosmid. There is some indication that they may be co-regulated. Therefore aberrant expression of PP2C ⁇ can be expressed as behavioral changes.
  • the levels of PP2C ⁇ activity are extremely high in cardiac and kidney tissues compared to other tissues. Therefore alterations in PP2C ⁇ activity will be reflected in these tissues.
  • the present invention provides a method of suppressing gene amplification by interrupting the binding or action of DNA polymerase a primase and RNA polymerase II with the gene product of PP2C ⁇ by preparing an antisense vector which will specifically target the binding region of DNA polymerase ⁇ primase and RNA polymerase II to the PP2C ⁇ gene product and delivering the vector to the cells as based on the observations set forth in Example 9.
  • Applicants have observed that in tumor cells pp2c ⁇ binds to the CTD domain of RNA polymerase II. Therefore alternatively, delivery of a peptide with the CTD domain can be used via competitive binding strategies to control the binding leading to gene amplification.
  • CO60 is a cell line of SV40 transformed Chinese hamster embryo cell lines [Lavi, 1981].
  • the OD cell line was established by transfection of Chinese hamster embryonic cells with origin deleted SV40 DNA [Lavi, 1985].
  • the mouse DA3 cell line was derived from mammary tumors syngeneic to BALB/c mice [Sotomayor et al, 1991].
  • JDT277 virus contains the portion of the AAV2 genome, which encodes the viral Rep proteins, the AAV terminal inverted repeats (TIRs) and the prokaryotic neomycin phosphotransferase gene (neo), conferring resistance to G418.
  • the neo gene was inserted at nucleotide 1882, resulting in carboxy terminus truncated Rep proteins. The truncation of the rep proteins does not affect the ability of the AAV/neo virus to replicate in Adenovirus coinfected human cells.
  • a characteristic trait of tumor cells is their capability to amplify DNA.
  • CO60 cells are used as a model system to study gene amplification and SV40 amplification can be induced in the cells as a results of treatment with carcinogens [Lavi, 1981; Aladjem and Lavi, 1992].
  • the cells were incapable to amplify SV40.
  • Most AAV/neo cell lines derived from CO60 cells lost their capability to amplify SV40 upon treatment with carcinogen in contrast to the parental CO60 cells [Tal Burstyn, 1993]. Extracts from AAV/neo cells derived from both OD4 and CO60 cells lost their capability to amplify SV40 in vitro [Winocour et al., 1992; Tal Burstyn, 1993].
  • a substantiated fraction of the cells displayed apoptotic nuclei showing condensed chromatin upon staining with acridine orange.
  • the cells displayed a strong shrinkage of the cytoplasm. Often the nuclei were disrupted into a multitude of micronuclei. These cells underwent apoptosis without losing their membrane integrity. EtBr did not penetrate into these cells, thus the cells were still alive.
  • a large amount of living apoptotic nuclei were found in the treated AAV positive cells compared to a considerably lower percentage in the treated (7.5 ⁇ g/ml MNNG) and control CO60 cells. The same pattern of staining repeated in all the AAV/neo cell lines. Hence, this apoptotic phenotype was a common feature to all the AAV/neo cells.
  • TDT terminal deoxynucleotidyl transferase
  • Applicants could detect a distinct pattern of nuclear staining, directly correlated to the typical degradation of chromatin in apoptotic cells. Since this reaction is specific, only the apopotic nuclei are stained. As a positive control for the efficiency of the technique, Applicants used CO60 nuclei treated with DNase.
  • Untreated AAV/neo cells and control and treated CO60 did not show any sign of fluorescence. This pattern of nuclear degradation appeared in all AAV/neo cell lines tested (approximately 20), however, the extent of fragmentation varied in the different lines.
  • the revertant cells designated C9-3-2 and C9-3-12, which were selected on the basis of loss of resistance to G418, and lost their integrated AAV sequences [Burstyn, 1993], still maintained their apoptotic phenotype following treatment with 2.5 ⁇ g/ml or 5 ⁇ g/ml MNNG.
  • Applicant focused on the analysis of one Chinese hamster cell lines, 9-3, derived from CO60 cells (FIG. 3).
  • the integrated AAV undergoes duplication in this cell line and the chromosome harboring the AAV contains two regions in which AAV is integrated.
  • This duplication of AAV probably resulted from the massive rearrangement which occurred in the Chinese hamster genome following AAV integration.
  • the chromosome harboring the integrated AAV was altered and was different in many respects from all the typical Chinese hamster chromosomes, thus the identity of the chromosome could not be established.
  • FIG. 3 The integrated AAV and flanking cellular sequences for 9-3 were cloned into a phage (FIG. 3).
  • the viral genome underwent several changes. Sequences downstream to the AAV p5 promoter were deleted and replaced by a cellular fragment “CHINT”. In addition, deletions and rearrangements in the 5′ portion of the AAV/neo genome were observed. In contrast, the region coding for the Neo gene and the 3′ end of the viral genome remained intact. (Similar alterations were observed in all AAV/neo Chinese hamster and mouse cell lines tested).
  • MMDA cosmid MMDA (Access #M63796) which was automatically sequenced and contained PP2C ⁇ first coding exon in position 59770 in MMDBC, GenBank accession #M89657, as well as 35-3.seg and 35-T7 as shown in FIG. 3.
  • MMDA and MMDBC are two cosmids in the same contig. (More details on the sequences are found in Example 10 herein below).
  • pp2c ⁇ might be a cell marker itself.
  • the prior art does not provide information about pp2c ⁇ expression in tumor cells.
  • pp2c ⁇ might have a role during myogenic differentiation. [Ohishi, 1992].
  • pp2c ⁇ Based on the presence of a 10 amino acid motif which appears also in other transcription factors, pp2c ⁇ might function like a transcription factor and might regulate transcription in the cell under specific growth conditions and tissues. It can thus behave like E2F which is a major transcription factor and can act when improperly expressed either as an oncogene or as a tumor suppressor factor [Weinberg, 1996]
  • PP2C ⁇ appears to be important in development.
  • IRES internal ribosome entry site
  • the findings by other laboratories that AAV infection effects specifically tumor cells might have two explanations: 1) The virus does not infect normal cells or cannot integrate into their genome in a specific manner. 2) Alternatively, if AAV integrates into PP2C ⁇ in normal cells the disruption of this gene might not effect them or might be lethal not allowing the survival of such cells.
  • CEA cancer embryonic antigen
  • the steps of the method are: (1) binding of antigen to a solid phase; (2) binding of the antibody to the antigen; and (3) binding of a labeled secondary antibody to the complex.
  • binding constant amounts of rpp2c ⁇ to the solid phase Applicants have used this technique to detect and quantitate monoclonal antibodies during the rounds of cloning, and to compare polyclonal antibodies from different rabbits and bleedings.
  • the assay has also been used to detect pp2c ⁇ in crude extracts of tissues and cell cultures.
  • the steps of the method are: (1) preparation of antigen sample; tissue extracts, cell culture extracts or rpp2c ⁇ preparations; (2) resolution of the sample by SDS-PAGE; (3) transfer of the separated proteins to a nitrocellulose membrane; (4) blocking nonspecific sites on the membrane; (5) incubation with poly- or monoclonal antibody; and (6) detection by labeled secondary antibody.
  • Applicants have used immunoblotting for characterization of antibodies described herein above and for detection of pp2c ⁇ in cell and tissue extracts. [Harlow and Lane]
  • the method steps are: (1) immobilization of monoclonal antibodies to a solid matrix (anti-mouse IgG conjugated agarose); (2) binding of antigen to immobilized antibodies; (3) resolution of bound proteins on SDS-PAGE; and (4) immunoblotting and detection of antigen by affinity purified rabbit polyclonal antibodies.
  • the method has been used to estimate the amount and the molecular mass of different sized pp2c ⁇ and ⁇ polypeptides that were discovered.
  • PP2C ⁇ gene product is purified from the mouse cells by general procedure, and its activity is assayed by its ability to dephosphorylate [32P] casein [McGowan and Cohen, 1988].
  • Rat PP2C- ⁇ cDNA specific primers were used for reverse transcription and PCR. These primers were obtained from General Biotechnology, Rehovot, and used without further purification. The primers' position is according to the rat kidney nucleotide sequence of PP2C ⁇ cDNA reported by Tamura et al., [1989] in the Genbank (accession number: Gb_ro: Ratpp2c, J04503).
  • Antisense RNA [0131]
  • the differential display method is used [Liang and Pardee, 1992; McClelliand et al., 1995]. This method is directed toward the identification of differentially expressed genes among approximately 15,000 individual mRNA species in a pair of mammalian cell population such as infected and uninfected cells, and recovering their cDNA and genomic clones.
  • the strategy of the method consists of the following steps: (1) Reverse transcription in fractions using a set of anchored primers, (2) amplification of cDNA species from each fraction using a set of arbitrary primers and anchored primers by labeled PCR, (3) electrophoretic separation of the resulting fragments on sequencing gel, (4) reamplification of fragments that are different in the two situations, cloning and sequencing, and (5) confirmation of differential expression by an independent RNA analysis technique.
  • RNA is isolated from cells as described by Sambrook et al.
  • the RNA is reverse transcribed with an oligo dT primer designed to bind to the 5′ boundary of the poly A tail.
  • the cDNA is amplified in a PCR reaction with the oligo dT primer and a second 10-mer arbitrary in sequence. 40 cycles of PCR are done in the presence of [35S]-dATP, in the following conditions: 94° C. for 30 seconds, 42° C. for 60 seconds and 72° C. for 30 seconds.
  • the amplified cDNAs are separated on a 6% sequencing gel, then exposed to X-ray film.
  • Bands of interest are cut out from the gel, and reamplified with the same primers as used to generate the original PCR product.
  • Northern blot hybridizations is performed. Fragments of interest are cloned using a TA cloning Kit, and sequenced. Genes detected by this method are hybridized to Northern blots from the appropriate cells.
  • Chromatin is partially purified and digested by micrococcal nuclease [Roth et al., 1990]. Purified DNA fragments are digested with a unique restriction enzyme to generate a series of fragments with one end defined by micrococcal nuclease and the other defined by the restriction enzyme. Fragments are separated by agarose gel electrophoresis, transferred to nitrocellulose filters and probed with labeled DNA fragments. Naked DNA is purified and processed similarly. Nucleosome position and nuclease sensitive regions are inferred by comparison of fragments from naked DNA and chromatin.
  • the methylation state of genes can indicate chromatin changes.
  • Gene specific DNA methylation is measured by the methylation assay [Kafri et al., 1992]. In this method, total cellular DNA is digested with methyl-sensitive enzymes, such as HpalI or KhaI, and specific fragments of DNA that contain these sites are amplified by flanking oligonucleotide primers. If a specific site is methylated, the amplification will proceed normally. On the other hand, the presence of an unmethylated site will result in digestion of the fragment and the subsequent failure to visualize the amplification product. When properly calibrated, this assay is linear over a wide range of DNA concentrations and can be used to accurately measure the degree of DNA methylation at specific sites.
  • DA3 cells were infected with the JDT277 AAV/neo hybrid virus according to Winocour et. al. [1992], with slight modifications. Single colonies were isolated and amplified by serial passages in the presence of the antibiotic G418. The resistant cell lines were designated DA3J.
  • the DA3 cell line was derived from the in vivo D1-DMBA-3 mammary tumor syngeneic to BALB/c mice.
  • the DA3 cell line produces tumors in BALB/c mice with the same growth kinetics and expresses the same tumor associated antigen (Ag) on its surface as the parental tumor.
  • the cells express specific markers for tumor cells, and cease to express specific Ag typical to normal breast cells [Sotomayor et al., 1991].
  • JDT277 contains the portion of the AAV2 genome, which encodes the Rep proteins, the AAV terminal inverted repeats (TIRs) and the prokaryotic neomycin phosphotransferase gene (neo), conferring resistance to G418.
  • the neo gene was inserted at nucleotide 1882, resulting in carboxy terminus truncated Rep proteins. The truncation of the Rep proteins does not affect the ability of the AAV/neo virus to replicate in Adeonvirus coinfected human cells.
  • Genomic DNA isolated from DA3J1-DA3J7 clones was digested with different restriction enzymes (BglII or EcoRI), electrophoresed and hybridized to radiolabelled AAV DNA.
  • the hybridization pattern is different in each clone, probably due to rearrangement of the AAV genome. Indeed it is known that integration of AAV DNA is frequently accompanied by alterations within the viral sequences [Walz and Schlehofer, 1992].
  • Genomic DNA from parental DA3 and DA3J clones was digested with BglII or EcoRI. Following electrophoresis the blots were hybridized once with the cellular sequence from the virus/cell junction, isolated from C9-3 (psL9-6), and once with radiolabelled AAV DNA. In three of the cell lines (DA3J3, DA3J4 and DA3J6) the cellular probe and the AAV probe hybridized to common bands. Using PP2C ⁇ probe applicant found in BglII digested DNA that both AAV and PP2C ⁇ hybridized to the same bands.
  • the plating efficiency of the DA3J cells was reduced compared to the plating efficiency of the parental DA3 cells, by 11% (DA3J2) to 54% (DA3J3).
  • the DA3J cells show increased sensitivity to UV irradiation compared to the parental DA3 cells. There is a decrease of 5% to 55% in the survival rate of the DA3J cells compared to the DA3 cells.
  • DA3J3 shows the lowest plating efficiency, and the highest sensitivity to UV irradiation. This may be due to the fact that DA3J3 contains two integrated AAV molecules, while DA3J1 and DA3J2, contains only one.
  • Protein phosphatase activity was found in crude extracts of cultures harboring the recombinant plasmid, as measured by the method of McGowan and Cohen [Methods Enzymol. 159: 416-429, 1988].
  • rpp2c ⁇ Purified rpp2c ⁇ was used for monoclonal antibody preparation, by mouse hybridoma production as described herein above. Hybridoma colonies were screened by antibody capture assay (see herein below) and by immunofluorescent cell staining. Positive colonies were subjected to two rounds of cloning and screening by the same methods. Finally, eight (8) positive clones were chosen for further study. Antibodies from these clones were collected as tissue culture supernatants and also as ascitic fluid.
  • the antibody was raised in rabbits and was affinity purified against the rpp2c ⁇ . This antibody was used in most of the histochemical analyses.
  • a rabbit polyclonal antibody raised against PNKDNDGGA (SEQ ID No:3), the carboxy terminal of pp2c ⁇ .
  • Table 4 provides the characterization of eight monoclonal antibodies by antibody capture assay, immunoblotting and immunoprecipitation. The combination of these assays allow the isolation of monoclonal antibodies with the proper specificity of the present invention.
  • Paraffin blocks obtained from normal breast and breast carcinoma were stained with 801 antibodies specific to pp2c ⁇ and then with secondary antibodies coupled to peroxidase.
  • the substrate was DAB.
  • the samples were counterstained with methylene blue.
  • the antibodies used were monoclonal 2A3 which are specific to pp2c ⁇ .
  • the magnification was ⁇ 400. Normal liver and hepatoma tissue and normal colon and colon carcinoma tissue was also tested.
  • pp2c ⁇ protein phosphatase 2C ⁇
  • CHE Chinese hamster embryo
  • CO60 non permissive SV40 transformed Chinese hamster cells
  • RNA samples were denatured at 65° C. for 10 minutes in the presence of 0.5 M oligo dT (15 mer) as an anti-sense primer, and immediately chilled on ice.
  • First strand cDNAs were obtained after 60 minutes at 37° C. in a 50 ⁇ l reaction mixture containing: 0.25 mM dNTPs (Promega), 10 mM DTT, 20u RNasin, 50u MMLV reverse transcriptase and 5 ⁇ l of 10 ⁇ reaction buffer (STRATAGENE). Following inactivation at 95° C.
  • This system relies on constitutive expression of a tetracycline-controlled transactivator (tTA) fusion protein which combines the tetracycline repressor with the activating domain of herpes simplex VP16.
  • the tTA was constitutively expressed in rat fibroblasts and in HeLa cells. In these two cell lines the tTA stimulates transcription from a minimal promoter derived from the human cytomegaloviruspromoter and the tetracycline operator. Upon addition of tetracycline the stimulation of transcription by tTA is inhibited.
  • Clones were prepared by stable transfection of the two cell lines with expression vectors that contain the PP2C ⁇ mRNA in the sense and in the antisense orientation, under the control of the tTA-dependent promoter.
  • the DNA fragment coding for the rat PP2C ⁇ mRNA was prepared by thermal cycling amplification.
  • the template for the amplification reaction was the insert of plasmid skPP2C (PP2C ⁇ cDNA cloned into the sk BLUESCRIPT plasmid).
  • the upper primer used in the amplification reaction contains the sequence coding for the first six amino acids of the rat PP2C ⁇ (Met Gly Ala Phe Leu Asp; SEQ ID No:9).
  • the sequence of the upper primer is the following: 5′ CGGGATCCGC ATGGGAGCAT TTTTAGAC 3′ (SEQ ID No:10).
  • the lower primer used in the amplification reaction contains the sequence coding for the last five amino acids and the stop codon of the rat PP2C ⁇ (Thr Asp Asp Met Trp ***; SEQ ID No:11).
  • the sequence of the lower primer is the following: 5′ CGCGGATCCT TACCACATAT CATCAGT 3′ (SEQ ID No:12).
  • the orientation of the cDNA insert with respect to the promoter was determined by restriction map analysis. Plasmids that contain the cDNA in the sense orientation (pYM001) and in the antisense orientation (pYM002) were selected (FIG. 9).
  • the sequence of the DNA insert of plasmid pYM001 was determined by automatic DNA sequence analysis.
  • the primers used for sequencing analysis were the same as the one used for cloning.
  • the results of this analysis show that the sequence of the cloned fragment is identical to that of rat PP2C ⁇ and that no mutation was introduced during the amplification reaction.
  • Plasmids pYM001 and pYM002 were introduced in the rat fibroblast and in the HeLa cell lines which constitutively express the tTA, by CaPO 4 coprecipitation with plasmid pBSpac. Plasmid pBSpac contains a genetic selective marker, that confers puromycin resistance.
  • the integrated AAV in SV40 transformed Chinese hamster cells (line 9-3 and other cell lines) is responsible for the suppression of the carcinogen induced SV40 amplification.
  • the viral element responsible for the suppression of SV40 amplification (silencer, SEQ ID No:13) was defined using a transient assay for SV40 replication [Yang et al, 1995] to demonstrate that the AAV rep protein is responsible for the suppression of SV40 replication.
  • This assay is based on transfection of the human kidney cell line 293 with an SV40 vector containing the coding region for T antigen and the viral origin of replication and cotransection with several different constructs derived from the vicinity of AAV integration in 9-3, SV40 transformed Chinese hamster embryo cells containing an integrated AAV and DA357 (a mouse cell line harboring the integrated AAV in which the transformed phenotype was altered following AAV integration).
  • This element was termed SV40 silencer (SEQ ID No:13) though in an alternative embodiment only 21 nucleotides, 125-145, (SEQ ID No:14) are responsible.
  • the blot provides the following:
  • Lane 1 Cotransfection with pSK1 and pAV2 (a plasmid containing the whole AAV genome and expressing the rep protein). Note that SV40 replication is suppressed.
  • Lane 2 Transfection with the SV40 plasmid pSK1 SV40 replicates. Replication of the SV40 template is observed.
  • Lane 3 Cotransfection of pSVK1 and a plasmid harboring 138 bp from the AAV genome (nucleotides 125-263). There was a suppression of SV40 replication by this element.
  • Lane 4 Cotransfection of pSVKl with pSL9-6 (non AAV DNA sequences).
  • suppression of SV40 replication can be obtained in 293 cells by Rep expression and by the 64 bp silencer element in a transient assay.
  • the 21 bp (mini-silencer) from AAV genome modulates PP2C ⁇ activity as well by interaction and activation of a control region.
  • the silencer can act as a dominant negative element interacting directly and/or indirectly with proteins associated with the replication of SV40.
  • PP2C can regulate the action of such proteins by dephosphorylation.
  • An example for such interplay can be the DNA polymerase ⁇ primase. It is possible that the rep protein is directly also involved in such interactions.
  • dephosphorylate DNA polymerase a primase is responsible for the initiation of SV40 DNA replication during the carcinogen induced amplification in CO60 cells. Moreover that this phosphorylate—dephosphorylate process are controlled by the cell cycle. Thus PP2C ⁇ can modulate the activity of the DNA polymerase ⁇ primase depletion of PP2C ⁇ due to its binding to rep directly or indirectly might lead to aberrant phosphorylic of the DNA polymerase ⁇ primase and to its failure to initiate SV40 DNA replication.
  • liver extracts were immunoprecipitated with the different monoclonal antibodies raised against rpp2c ⁇ (see Example 3 herein above). The precipitates were divided into two aliquots which were separated on 12% PAGE and blotted. Each set of immunoprecipitates was challenged with the following polyclonal antibodies:
  • FIG. 13 Norther blot analysis (FIG. 13) of mRNA derived from several tissues displayed several bands of RNA which hybridized with probes derived from the 5′UTR of pp2c ⁇ or with the entire PP2C ⁇ cDNA probe. The RNA was extracted from different tissues and different sizes of RNA appeared in these tissues.
  • Table 6 summarizes the protein or RNA sequence homology that was found for the 10 amino acid pp2c ⁇ carboxy terminal peptide: NDDTDSASTD (SEQ ID No:1). This peptide was used to raise polyclonal antibody 801 as described herein above.
  • RNA polymerase II The carboxy cellular domain (CTD) of the RNA polymerase II fused to GST and bound the fusion complex to sepharose gluthation beads and mixed with HeLa cell extract. Following PAGE and blot the proteins bound to the carboxy terminal domain (CTD) of the RNA polymerase bound also to pp2c ⁇ . Both 801 and 2A3 were used with the blot. The size of the associated pp2c ⁇ was approximately 43 kD. Thus, RNA polymerase II is associated with pp2c ⁇ .
  • pp2c ⁇ dephosphorylates RNA polymerase II and thus regulates the initiation of mRNA synthesis on specific messenger. This peptide can be used to control and regulate transcription facilitated by other factors.
  • Two ⁇ clones containing the AAV integration site were prepared. (1) One was derived from the chinese hamster cell CO60 designated ⁇ SL9-1 (schematic diagram in FIG. 3; SEQ ID Nos:15 and 16). Parts were sequences as indicated in FIG. 3. A further sequence AN8T7 (SEQ ID No:18) was derived from plasmid pSL9-8 (FIG. 3). (2) The second ⁇ phage was cloned from the cell line DA3J7 and was not mapped. Portions were subcloned to plasmids and part sequenced as set forth in 5h-1 (SEQ ID No:17) A sequence comparison shows that 5h-1 is homologous to AN8T7.
  • TPD1 of Saccharomyces cerevisiae encodes a protein phosphatase 2C-like activity implicated in tRNA splicing and cell separation. Mol. Cell. Biol. 14:3634-3645.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • General Health & Medical Sciences (AREA)
  • Biochemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Medicinal Chemistry (AREA)
  • Immunology (AREA)
  • Biophysics (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Analytical Chemistry (AREA)
  • Microbiology (AREA)
  • Biotechnology (AREA)
  • General Engineering & Computer Science (AREA)
  • Pathology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Oncology (AREA)
  • Physics & Mathematics (AREA)
  • Biomedical Technology (AREA)
  • Virology (AREA)
  • Hospice & Palliative Care (AREA)
  • Pharmacology & Pharmacy (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Investigating Or Analysing Biological Materials (AREA)
US09/029,479 1995-09-01 1996-08-30 Manipulation and detection of protein phosphatase 2c-pp2calpha - expression in tumor cells for cancer therapy, prevention and detection Abandoned US20030099611A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US10/678,477 US20040197798A1 (en) 1995-09-01 2003-10-03 Manipulation and detection of protein phosphatase 2C - PP2Calpha - expression in tumor cells for cancer therapy, prevention and detection

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US311495P 1995-09-01 1995-09-01
CN96196623A CN1194667A (zh) 1995-09-01 1996-08-30 用于癌症治疗、预防和检测的蛋白质磷酸酶2C-PP2Cα-在肿瘤细胞中表达的操作和检测方法
PCT/IB1996/001021 WO1997010796A2 (en) 1995-09-01 1996-08-30 MANIPULATION AND DETECTION OF PROTEIN PHOSPHATASE 2C - PP2Cα - EXPRESSION IN TUMOR CELLS FOR CANCER THERAPY, PREVENTION AND DETECTION

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB1996/001021 A-371-Of-International WO1997010796A2 (en) 1995-09-01 1996-08-30 MANIPULATION AND DETECTION OF PROTEIN PHOSPHATASE 2C - PP2Cα - EXPRESSION IN TUMOR CELLS FOR CANCER THERAPY, PREVENTION AND DETECTION

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US10/678,477 Division US20040197798A1 (en) 1995-09-01 2003-10-03 Manipulation and detection of protein phosphatase 2C - PP2Calpha - expression in tumor cells for cancer therapy, prevention and detection

Publications (1)

Publication Number Publication Date
US20030099611A1 true US20030099611A1 (en) 2003-05-29

Family

ID=27664236

Family Applications (1)

Application Number Title Priority Date Filing Date
US09/029,479 Abandoned US20030099611A1 (en) 1995-09-01 1996-08-30 Manipulation and detection of protein phosphatase 2c-pp2calpha - expression in tumor cells for cancer therapy, prevention and detection

Country Status (7)

Country Link
US (1) US20030099611A1 (ja)
EP (1) EP0876507A4 (ja)
JP (1) JPH11512294A (ja)
CN (1) CN1194667A (ja)
AU (1) AU723055B2 (ja)
IL (1) IL123475A0 (ja)
WO (1) WO1997010796A2 (ja)

Families Citing this family (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0874052A3 (en) * 1997-04-22 1999-02-24 BIOPHARM GESELLSCHAFT ZUR BIOTECHNOLOGISCHEN ENTWICKLUNG VON PHARMAKA mbH Nucleic acid encoding a human protein phosphatase
US5853997A (en) 1997-06-11 1998-12-29 Incyte Pharmaceuticals, Inc. Human protein phosphatase
US5948902A (en) * 1997-11-20 1999-09-07 South Alabama Medical Science Foundation Antisense oligonucleotides to human serine/threonine protein phosphatase genes
JP5734978B2 (ja) * 2009-08-19 2015-06-17 メルク パテント ゲゼルシャフト ミット ベシュレンクテル ハフツングMerck Patent Gesellschaft mit beschraenkter Haftung Ffpe材料におけるインテグリン複合体検出のための抗体
CN113527289A (zh) * 2015-09-23 2021-10-22 米纳瓦生物技术公司 筛选分化干细胞的试剂的方法
JP2022537581A (ja) * 2019-06-21 2022-08-26 クラリス コーポレーション Ppm1阻害剤およびその使用方法
CN111534517A (zh) * 2020-05-16 2020-08-14 扬州大学 一种抑制原癌基因c-myc表达的反义RNA MYC-AS1及其应用
WO2024076285A1 (en) * 2022-10-05 2024-04-11 Ivarsson Ylva Peptide targeting sars-cov-2 nsp9

Also Published As

Publication number Publication date
EP0876507A2 (en) 1998-11-11
WO1997010796A2 (en) 1997-03-27
JPH11512294A (ja) 1999-10-26
AU6998096A (en) 1997-04-09
EP0876507A4 (en) 2003-07-30
WO1997010796A3 (en) 1997-06-19
IL123475A0 (en) 1998-09-24
CN1194667A (zh) 1998-09-30
AU723055B2 (en) 2000-08-17

Similar Documents

Publication Publication Date Title
US8735066B2 (en) Tumor suppressor designated TS10Q23.3
US5801236A (en) Probes for MTS1 gene and polynucleotides encoding mutant MTS1 genes
JP4768783B2 (ja) Ts10q23.3と称する腫瘍抑制因子
JPH08509504A (ja) プロテインキナーゼ
JP2010051330A (ja) Ts10q23.3と称する腫瘍抑制因子
AU723055B2 (en) Manipulation and detection of protein phosphatase 2C - PP2Calpha - expression in tumor cells for cancer therapy, prevention and detection
US20040197798A1 (en) Manipulation and detection of protein phosphatase 2C - PP2Calpha - expression in tumor cells for cancer therapy, prevention and detection
CA2253433A1 (en) Mammalian regulator of nonsense-mediated rna decay
US5843756A (en) Mouse MTSI gene
US20020142417A1 (en) Antioxidant protein 2, gene and methods of use therefor
US20020098174A1 (en) 33166, a human hydrolase-like molecule and uses thereof
CA2244744A1 (en) Lyst1 and lyst2 gene compositions and methods of use
CZ254797A3 (cs) Kinasa příbuzná PIK kontrolního prvku buněčného cyklu, látky a způsoby
JP2002515732A (ja) 細胞周期チェックポイントpik関連キナーゼ物質及び方法

Legal Events

Date Code Title Description
AS Assignment

Owner name: RAMOT-UNIVERSITY AUTHORITY FOR APPLIED RESEARCH AN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:LAVI, SARA;REEL/FRAME:010453/0131

Effective date: 19990607

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION